Location via proxy:   [ UP ]  
[Report a bug]   [Manage cookies]                

The Future of Recombinant Host Defense Peptides

Download as pdf or txt
Download as pdf or txt
You are on page 1of 16

Roca‑Pinilla et al.

Microbial Cell Factories


https://doi.org/10.1186/s12934-022-01991-2
(2022) 21:267
Microbial Cell Factories

REVIEW Open Access

The future of recombinant host defense


peptides
Ramon Roca‑Pinilla1, Leszek Lisowski1,2, Anna Arís3* and Elena Garcia‑Fruitós3*

Abstract
The antimicrobial resistance crisis calls for the discovery and production of new antimicrobials. Host defense peptides
(HDPs) are small proteins with potent antibacterial and immunomodulatory activities that are attractive for transla‑
tional applications, with several already under clinical trials. Traditionally, antimicrobial peptides have been produced
by chemical synthesis, which is expensive and requires the use of toxic reagents, hindering the large-scale develop‑
ment of HDPs. Alternatively, HDPs can be produced recombinantly to overcome these limitations. Their antimicrobial
nature, however, can make them toxic to the hosts of recombinant production. In this review we explore the different
strategies that are used to fine-tune their activities, bioengineer them, and optimize the recombinant production of
HDPs in various cell factories.
Keywords: Host defense peptides, Antimicrobial proteins, Antimicrobial resistance, Inclusion bodies, Recombinant
production

Background resist bacterial infections thanks to the action of cecropin


In 2019 alone, 1.27 million people died globally due to [5]. Another example is the potent antimicrobial activ-
antimicrobial-resistant bacteria (ARB) [1]. At the current ity of rabbit neutrophils due to defensins [6] and the skin
rate of resistance development, 10 million people will die wound-healing abilities of an African clawed frog, thanks
by 2050 due to our inability to treat infections [1]. There to secreted magainins [7]. Since then, the field of HDPs
is, therefore, a severe need to find suitable alternatives as exploded and today there are more than 3000 known
effective as conventional antibiotics or that can be used peptide sequences that come from all domains of life,
in a combinatorial treatment [2]. including HDPs [8].
One potential alternative is the use of host defense The characteristics that usually define HDPs are their
peptides (HDPs), which are a diverse and well-studied short amino acidic sequences (between 12 and 50 amino
class of bioactive peptides (AMPs) that all multicellu- acids) [4], a net positive charge [5], a certain degree of
lar organisms produce as a defense mechanism against hydrophobicity [9] and a wide range of broad-spectrum
pathogenic microbes [3, 4]. HDPs were discovered in the biological activities [10]. Among these activities, HDPs
1980s thanks to the keen eye of researchers that could have microbicidal (effective against bacteria, virus, and
not explain what they observed with their current under- fungi) [11–13], antibiofilm [14, 15], and immunomodula-
standing of immunity. For instance, Cecropia moth pupa tory activities [16–18]. Interestingly, it might be difficult
that lacked antibodies or lymphocytes were still able to for microorganisms to develop resistance against HDPs
because of their multiple modes of action, which may
ultimately lead to microbial death [3].
*Correspondence: anna.aris@irta.cat; elena.garcia@irta.cat Their well-known characteristics make them ame-
3
Department of Ruminant Production, Institut de Recerca i Tecnologia nable to engineering [19–21] (Fig. 1A, B), peptide
Agroalimentàries IRTA​, 08140 Caldes de Montbui, Spain repurposing, such as engineered venoms that can be
Full list of author information is available at the end of the article modified to become non-toxic HDPs [22] (Fig. 1A),

© The Author(s) 2022. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which
permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the
original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or
other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line
to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory
regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this
licence, visit http://​creat​iveco​mmons.​org/​licen​ses/​by/4.​0/. The Creative Commons Public Domain Dedication waiver (http://​creat​iveco​
mmons.​org/​publi​cdoma​in/​zero/1.​0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


Roca‑Pinilla et al. Microbial Cell Factories (2022) 21:267 Page 2 of 15

Fig. 1 Schematic figure showing several strategies that can be used to engineer antimicrobial peptides. A Rational modification of peptides to
improve some of their features; B multidomain proteins based on the combination of different HDPs; C design new peptides based on their known
properties; or D find encrypted peptides “buried” in known protein sequences

development of multidomain proteins based on the including potential toxicity, low stability, half-life, and a
combination of different HDPs (Fig. 1B), de novo high production cost.
designs [9, 23] (Fig. 1C), or aid in the discovery of Together with the need to be safe and effective, one of
hidden peptides within larger protein structures [24] the big questions that remains is how to produce them in
(Fig. 1D). All these new technologies yield an almost large quantities in a sustainable way and with a competi-
unlimited potential to modify known sequences or dis- tive price. Besides, some strategies including chemical
cover new peptides and modes of action. modifications and delivery vehicles are being studied to
There are multiple ways to classify HDPs, considering improve the properties of these peptides [28].
their secondary structure or common ancestry, for exam- The first HDPs were isolated and purified from their
ple. In vertebrates, there are two major families of HDPs: natural sources, but this process is difficult and time-
cathelicidins and defensins [10]. The latter have a com- consuming, and results in low yields, making it diffi-
mon β-sheet core stabilized by three disulphide bridges. cult to scale up (Fig. 2). Most of the studies to date use
Depending on how the cysteine residues link together, chemically synthesized HDPs [29], and, by automated
defensins are classified into α-, β-, and θ-defensins [10]. solid-phase peptide synthesis (SPPS) [29]. This strategy
Instead, over one third of the cathelicidins are α-helical. involves the repetitive binding of different amino acids to
Cathelicidins are produced as prepropeptides that need obtain the desired peptide sequence, which is bound to a
to be secreted and then cleaved by serine proteases [25]. resin support. Once the sequence is obtained, the peptide
However, there are other families, such as histatins, can be retrieved from the solid support with high purity
which are histidine rich HDPs from mammals’ saliva via a cleavable linker.
(Fig. 1D) [10, 24, 26]. Yet, in addition to the high production costs, one of
Although some peptide-based antimicrobials are in the main problems of chemical synthesis is its environ-
advanced clinical trials, none of them has been granted mental impact, due to the excessive use of organic sol-
regulatory approval [27]. There are many reasons, vents during the process [30]. Thus, it does not suit the
beyond the scope of this review, as to why that is the case, needs of large-scale production [31]. Besides, chemical

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


Roca‑Pinilla et al. Microbial Cell Factories (2022) 21:267 Page 3 of 15

Fig. 2 Advantages and disadvantages of different recombinant expressions systems to produce HDPs [52, 108–110]. Only the most utilized systems
are shown, as there are very few instances where mammalian cells have been used for recombinant HDP production, as it is probably too expensive
and unnecessary in many cases. The bulk of the work with plant systems is based on transgenic plants to enhance their properties with genetic
modifications, but not necessarily recombinant HDP production per se

synthesis can be tricky for longer peptide sequences Cell factories for recombinant HDP production
of more than 35 amino acid residues [32]. In this con- As an alternative to chemical synthesis, technologies
text, our goal in this review is to discuss the feasibil- based on recombinant DNA have been explored for the
ity of recombinant HDP production as an alternative to biosynthesis of HDPs. The recombinant production of
chemical synthesis, considering several of the microbial these peptides offers a more flexible, sustainable, scal-
cell factories that have been used so far, as well as vari- able, and cost-effective production [31]. Many organisms
ous protein forms and strategies that lead to success in can be used as hosts for recombinant HDP production,
recombinant HDP production endeavors. including plants, insect cells, mammalian cells, yeast,

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


Roca‑Pinilla et al. Microbial Cell Factories (2022) 21:267 Page 4 of 15

and bacterial cells [33]. However, choosing the optimal Exploiting all of these, a group from MIT [43] expressed
expression organism is critical to ensure proper protein apidaecin, an insect HDP, fused to human serum albumin
yields, biological function, and final cost (Fig. 2). (HSA) in the yeast Pichia pastoris. They obtained yields
of more than 700 mg/L with no cell lysis and no debris
Recombinant production in bacteria removal steps required. However, because it is a fusion
The most extensively used host for recombinant expres- construct, downstream cleaving of the fusion tag was
sion of proteins and peptides are bacteria, as they are necessary. Fish [44], human [45] and fungal [46] peptides
easy to manipulate, grow fast and use inexpensive media. have also successfully been produced recombinantly in P.
Nevertheless, bacteria, have a limited ability to make pastoris (Table 1), showing evidence that it can be a use-
disulphide-bonds, glycosylation, and other post transla- ful alternative to the most used expression system for
tional modifications (PTMS) [34–36]. These limitations, recombinant production (E. coli). Saccharmoyces cerevi-
however, might not be critical for HDP recombinant pro- siae is another yeast-based system that has been used for
duction [37]. In the absence of a strict need for PTMS, the expression of HDPs [47, 48], albeit less than P. pasto-
the heterologous expression in bacteria is a reasonable ris. The yields (mg/L) for HDPs expressed in yeast found
approach for their production with sufficient conforma- in the relevant literature are highly variable [39], between
tional and functional quality. less than 0.1 to up to 831 mg/L, suggesting the need to
As it occurs for many other recombinant proteins, the fine tune the expression system for each of the peptides.
most utilized bacterium for HDP production is E. coli
(Table 1), since it has been widely studied as a recombi- Recombinant production in fungi
nant host, with an extensive knowledge of its genetics, The use of filamentous fungi for HDP production is still
biochemistry, and physiology [38]. There are well-estab- in its infancy. However, interest in their use is growing
lished protocols and a large catalogue of expression vec- due to their successful application in the recombinant
tors, and besides, it grows fast. In addition, some HDPs production of non-antimicrobial proteins, their abil-
have been successfully expressed in Lactococcus lactis ity to perform PTMS, scale-up ability, and inexpensive-
(Table 1) and Bacillus subtilis. Although there are only ness of culture. The fungal defensin plectasin, developed
few examples of HDPs produced in these LPS-free bac- for the treatment of Gram-positive bacterial infections
teria, they are appealing candidates considering their is produced recombinantly using Aspergillus oryzae as a
status as generally regarded as safe (GRAS). Most HDPs high efficiency recombinant protein expression system,
expressed in bacteria are produced with inducible expres- produced as a secreted product (Table 1) [49]. Another
sion systems yielding quantities that range between 2 and example is the recombinant production of a hybrid HDP
600 mg/L [39]. of magainin II-cecropin B, successfully expressed in
Cordyceps militaris with a yield of 3.86 mg/g of mycelium
Recombinant production in yeasts (Table 1) [50].
Sometimes, the production of cysteine rich cationic
HDPs fails in bacterial systems such as E. coli, due to an Recombinant production in insect cells
inefficient formation of disulphide bridges that leads to The first isolated HDP was cecropin (1980), an insect
improper folding and lack of bioactivity [40]. Another HDP [5]. Insect cell-based systems prove to be very valu-
concern when expressing HDPs in bacteria is their natu- able for the recombinant expression of HDPs. In gen-
ral lethality towards the host [41]. Therefore, switching to eral, the most prevalent systems are based on Drosophila
eukaryotic cells can be a suitable alternative to overcome melanogaster cell lines, although there is some work in
this challenge, allowing for the heterologous expres- mosquito and moth cells [51]. Two main approaches are
sion of HDPs. Yeasts are one of the simplest eukaryotic used to express HDPs in insect systems, either by using
organisms [42] and offer a good compromise between a Baculovirus gene expression system [35], or by stably
the higher complexities of eukaryotic cells and the rela- expressing a HDP by means of genome integration of the
tively simple and inexpensive recombinant production HDP-coding gene.
of prokaryotic systems (Fig. 2). Besides, they grow faster Even though they are more expensive than yeast and
than typical mammalian recombinant hosts such as prokaryotic-based systems, they offer PTMS such as gly-
Chinese Hamster Ovary (CHO) or Human Embryonic cosylation that might aid the proper structure and bioac-
Kidney (HEK293) cells. And yeasts are free from endo- tivity of HDPs [52], although it is not clear if PTMS are
toxins and harmful human viruses and can secrete large necessary in all instances, or at all, suggesting that their
amounts of heterologous recombinant proteins with lit- need (or their lack of ) might require to be studied on a
tle host cell protein secretion, which can simplify down- case-by-case basis. However, when the Baculovirus sys-
stream purification (Fig. 2). tem is used, the lytic cycle of the virus can cause large

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


Roca‑Pinilla et al. Microbial Cell Factories (2022) 21:267 Page 5 of 15

Table 1 Recombinant expression of HDPs and the different cell hosts used to express them
HDP Type Family Host Protein ­forma Fusion tag References

Apidaecin Insect AMP L. lactis subsp. cremoris Secreted – [111]


NZ9000
Apidaecin Insect AMP Pichia pastoris Secreted Human serum albumin [43]
(HSA)
Big defensins Molusc big defensin E. coli BL21(DE3) Solubilized – [112]
Bovine lingual antimicrobial Bovine defensin E. coli BL21(DE3) Soluble eGFP [37]
peptide (LAP) E. coli Origami B (DE3) Solubilized
Buforin II Histone H2A-derived AMP E. coli BL21 (DE3) Solubilized Cystein-rich acidic peptide [92]
(CAP)
Cathelicidin-BF (CBF) Cathelicidin B. subtilis WB800 N Secreted Intein [95, 157]
SUMO
CcDef2gene Insect defensin E. coli BL21(DE3) Solubilized 3xCcDef2 Casette [113]
Cecropin Cathelicidin P. pastoris Soluble Oleosin [114]
ChMAP-28, mini- Goat cathelicidins E. coli BL21(DE3) Solubilized Trx [115]
ChBac7.5Nα, and mini-
ChBac7.5Nα
CRAMP Cathelicidin E. coli BL21(DE3) Soluble SUMO [41]
Cryptidin-2 Defensin E. coli Rosetta-gami B (DE3) Soluble Trx [64]
E5 and E6 Bovine bactenecin deriva‑ E. coli BL21(DE3) Soluble SUMO [41]
tive
Egyptian maize defensin Plant defensin E. coli BL21(DE3) Soluble GST [116]
(MzDef )
FaAMP Fungal defensin E. coli BL21 (DE3) Soluble eGFP [117]
fBD Flounder defensin E. coli BL21(DE3) Soluble – [118]
Fowlicidin-1 Chicken cathelicidin E. coli BL21 (DE3) Soluble Calmodulin [119]
Fungal defensin-like pep‑ Fungal defensin P. pastoris Secreted – [120]
tide (DLP)
GL13K Encrypted ­peptide a E. coli BLR Soluble Elastin-like recombinamers [121]
Gloverin Insect antibacterial protein Drosophila melanogaster S2 Secreted – [52]
Human neutrophil peptide Human defensin P. pastoris Secreted Polyhedrin-eGFP [45]
1 (HNP1)
Human neutrophil pep‑ Human defensin E. coli strain Soluble – [122]
tide-1 (HNP-1) XPX-1
Human α-defensin 5 (HD5) Human defensin E. coli BL21(DE3) Solubilized eGFP [20, 21, 37]
E. coli Origami B (DE3) Trx
Human α-defensin 5 (HD5) Human defensin P. pastoris Secreted Alpha-factor [123]
Human β-defensin 1 (HBD1) Human defensin E. coli AD202 Solubilized C-terminal fragment of light [124, 125]
Soluble meromyosin (LMM)
Trx
Human β-defensin 1 (HBD1) Human defensin S. cerevisiae AH22 Secreted – [126]
Human β-defensin 118 Human defensin E, coli Rosetta (DE3) Soluble – [127]
Human β-defensin 2 (HBD2) Human defensin E. coli BL21(DE3) Soluble Trx [125, 128–131]
Solubilized Keto-steroid isomerase (KSI)
Glutathione-S-transferase
(GST)
Human β-defensin 3 (HBD3) Human defensin E. coli BL21(DE3) Soluble Trx [119, 132]
Calmodulin
Human β-defensin 4 (HBD4) Human defensin E. coli BL21(DE3) Soluble Trx [133]
Human β-defensin 6 (HD6) Human defensin E. coli Origami (DE3) pLys Soluble Trx [134, 135]
Human β-defensin DEFB136 Human defensin E. coli BL21(DE3) Soluble Intein-chitin binding [136]
domain (CBD)
Hybrid peptide Cecropin Cecropin B. subtilis WB800N Secreted Small ubiquitin modifier [137]
AD (SUMO)
IDR-1 Innate defense regulator E. coli BL21(DE3) Soluble SUMO [41]
(IDR)

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


Roca‑Pinilla et al. Microbial Cell Factories (2022) 21:267 Page 6 of 15

Table 1 (continued)
HDP Type Family Host Protein ­forma Fusion tag References

Indolicidin Cathelicidin E. coli BL21 (DE3) Soluble Calmodulin [119]


Insect defensin A Defensin S. cerevisiae Secreted Yeast pheromone mating [138]
factor α (MFα)
Lactoferrampin B Fragment of lactoferrin E. coli BL21 (DE3) Soluble Calmodulin [119]
LL-37 Cathelicidin E. coli BL21 (DE3) Soluble SmbP [41, 139]
Silk
SUMO
LsGRP1c Glycine Rich Protein from E. coli BL21 Soluble SUMO [140]
E. coli C41 (DE3)
E. coli C43 (DE3)
E. coli C41 (DE3) pLysS
E. coli C43 (DE3) pLysS
LvCrustinVII Crustacean AMP E. coli BL21(DE3) Solubilized – [141]
Magainin II F5W Cathelicidin E. coli BL21 (DE3) Soluble Calmodulin [119]
Magainin II-cecropin B Magainin/cathelicidin Cordyceps militaris Secreted Chimeric ­proteinb [50]
chimera hybrid
Melittin – E. coli BL21 (DE3) Soluble eGFP [117, 119]
Calmodulin
MIP-3α51-70 Chemokine fragment E. coli BL21 (DE3) Soluble Calmodulin [119]
MX226 Indolicidin derivative E. coli BL21(DE3) Soluble SUMO [41]
OrR214 and OrR935 Rice AMPs B. subtilis SCK Secreted – [142]
pBD-2-cecropin P1 chimera Defensin/cathelicidin B. subtilis Secreted Chimeric ­proteinb [143]
hybrid
Peptide P2 Designed peptide E. coli NM522 Solubilized Bovine prochymosin [144]
Pexiganan Magainin analogue E. coli BL21 (DE3) Soluble DAMP4 [145]
Pexiganan-honeybee Magainin analogue/silk- E. coli Rosetta 2 (DE3) Solubilized Silk [146]
silk chimera (modified fibre hybrid
magainin-2)
Plectasin Fungal defensin B. subtilis WB800N Secreted SUMO [147]
Plectasin Fungal defensin P. pastoris Secreted 4xPlectasin casette [46]
Porcine β-defensin 2 Porcine defensin E. coli BL21(DE3) Soluble – [141, 148]
(pBD-2)
PsDef5.1 Fungal defensin E. coli BL21(DE3) Codon‑ Soluble Thioredoxin (Trx) [149]
PlusRIL
Rosetta-gami 2(DE3)
Puroindoline A – E. coli BL21 (DE3) Soluble Calmodulin [119]
r(P)ApoBL, r(P)ApoBsa Encrypted ­peptidea E. coli BL21 (DE3) Solubilized Onconase [150]
rAvBD1-2–6–13 Chicken defensin L. lactis NZ3900 Soluble – [151]
Scorpine Defensin Anopheles gambie Secreted – [51]
Sericin-cecropin Silk-fibre/cathelicidin hybrid E. coli BL21 (DE3) Soluble Silk [152]
E. coli Rosetta (DE3)
Sesvania javanica defensin Defensin E. coli Origami 2 (DE3) Soluble Intein-CBD [153]
(Javanicin)
SMAP Cathelicidin E. coli BL21 (DE3) Soluble eGFP [117]
Snakin-1 (StSN1) Plant AMP Spodoptera frugiperda Secreted – [154]
(Baculovirus-infected insect
cells)
T9W Variant of pig myeloid anti‑ B. subtilis WB800N Secreted SUMO [155]
microbial peptide-36
Thanatin Insect AMP Human Embryonic Kidney Secreted – [156]
(HEK)293
Tilapia piscidin Piscidin P. pastoris Soluble – [44]
Tritrpticin Cathelicidin E. coli BL21 (DE3) Soluble Calmodulin [119]
Histidine tags were not considered to be fusion tags that help to express HDPs
Soluble: soluble protein produced in the cytoplasm; solubilized: protein solubilize from IBs; secreted: soluble protein secreted to the media
a
Denotes peptide fragment obtained from larger, non-antimicrobial proteins
b
We considered chimeric proteins those that do need cleaving of the tag as they add new and desired bioactivities

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


Roca‑Pinilla et al. Microbial Cell Factories (2022) 21:267 Page 7 of 15

amounts of recombinant peptide loss due to degradation activity while showing a sustained release and a reduc-
[53]. Yields of HDPs produced in insect systems are less tion in pro-inflammatory cytokines release when com-
well documented than for yeast or bacterial systems, and pared to the non-encapsulated peptide [62]. Another
range between 6 and 25 mg/L [39]. approach that has been studied is the use of Polyethyl-
ene glycol (PEG)-stabilized lipodisks to protect cationic
Forms of recombinant HDPs peptides [63]. There is also a recent study that evaluated
In general, recombinant HDPs have been produced in 3 the encapsulated form of recombinant HDPs. Kaur and
main forms (Table 1): soluble protein (secreted or intra- coauthors analyzed the PEGylated form of mouse alpha-
cellular), inclusion bodies (IBs) or as encapsulated solu- defensin cryptdin-2 [64] and they observed a two-fold
ble protein. decrease in the antimicrobial activity when cryptidin-2
is conjugated to PEG. This effect could be attributed to a
Soluble recombinant production masking effect of PEG, and further studies are needed to
Many HDPs have been successfully obtained through evaluate the impact of PEGylation size and site, as previ-
recombinant production in different expression sys- ously described [64]. Going a step further, Drayton and
tems in a soluble form (Table 1) [54, 55]. In E. coli and coworkers have designed an enzyme-cleavable HDP-PEG
L. lactis, HDPs are typically produced intracellularly and system for the delivery of active HDPs, which is based
purified after cell disruption (Table 1). In contrast, when on the release of the antimicrobial peptide at the site
B. subtilis, yeast or fungi are used, the sequences of the of infection after cleavage by a host enzyme [65]. Thus,
proteins of interest are, mostly, designed to be secreted although much remains to be done, the results obtained
to the growth media (Table 1). However, in many other so far are promising when it comes to increase peptide
cases, especially when using bacterial expression systems, stability and decrease some of the adverse effects.
HDPs aggregate forming IBs, being necessary to solubi-
lize these protein aggregates (Table 1), as detailed in “IBs Antimicrobial inclusion bodies
as a source of soluble HDPs” section. IBs are protein nanoparticles or aggregates whose forma-
Besides aggregation, another issue of recombinant bac- tion has been widely described in E. coli [56, 57], but also
terial expression of HDPs is their potential lethality to the in other microbial expression systems such as lactic acid
recombinant host due to their antibacterial nature [56, bacteria [66–70] and yeast [70, 71]. These aggregates can
57]. In addition, they are highly susceptible to proteolysis be easily purified [72] and offer interesting features not
due to their small size and positive charge. To overcome available in a soluble form. IBs are an active biomaterial
all these problems, the most common strategy is to pro- that has already been explored in several applications
duce soluble HDPs with a fusion partner or to produce such as cancer [73], biocatalysis [74], tissue regenera-
them as multidomain proteins, as described in detail in tion [75] and immunostimulation [76], as they are highly
“Strategies to optimize HDP production” section. stable protein nanoparticles with slow-release properties
[56, 57, 77]. Recently, two studies have proven that HDP-
Encapsulated soluble HDPs based IBs are biologically active against different patho-
Some groups have also been working on the develop- genic bacteria [20, 37]. In the first study, López-Cano
ment of HDP delivery systems to have a time-controlled et al. showed that human α-defensin 5 (HD5) and lingual
release to improve bioavailability and to minimize tox- antimicrobial peptide (LAP) IBs are highly active against
icity and proteolytic degradation and, in consequence, MRSA and Pseudomonas aeruginosa, with antimicrobial
increase stability, when administered in vivo [58]. Most activities comparable to the soluble counterpart [37].
of these studies, however, have been done using synthetic The second study showed the antibiofilm properties of
HDPs. For example, different HDPs have been encap- IB-decorated surfaces against a carbapenem resistant
sulated using polymeric lactic-co-glycolic acid (PLGA) Klebsiella pneumoniae [20], adding to the evidence that
nanoparticles, showing that the encapsulated peptide antimicrobial IBs can be effectively used against AMR
kept the antimicrobial activity and did not increase bacteria.
toxicity when compared to the naked peptide [59, 60]. To maintain antimicrobial activity, constant admin-
Other studies using PLGA microspheres decorated with istration of an antimicrobial that has a short half-life is
N-acetyl cysteine (NAC) for pulmonary drug delivery required, as concentrations under minimum inhibitory
have shown to have a good potential both in vitro and concentrations (MIC) will probably happen during treat-
in vivo [61]. Synthetic HDPs have been also nanoencap- ment, further increasing the appearance of AMRs. This is
sulated in lipid-based nanoparticles. A peptide from the why a slow-release profile seems to be vital to maintain
cathelicidin family was encapsulated in liposomes and constant antimicrobial levels for long periods, to get an
the authors proved that the peptide kept its antimicrobial optimal therapeutic benefit, where HDP-based IBs are

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


Roca‑Pinilla et al. Microbial Cell Factories (2022) 21:267 Page 8 of 15

promising protein format for antimicrobial applications denaturing protocols, this last strategy allows to obtain
[37]. Therefore, antimicrobial IBs can be used as nano- soluble protein from IBs without the need to use unfold-
pills that display antibacterial activity against different ing and refolding processes. Among these articles, some
AMR Gram-positive and Gram-negative strains. These of them have already successfully tried this approach
HDP-releasing nanopills can also be used to decorate using IBs formed by antimicrobial proteins [20, 21, 37].
plastic surfaces to avoid biofilm formation by bacteria Indeed, solubilized IB proteins show antimicrobial activ-
[20]. In addition, the IB format per se can be antimicro- ity against E. coli in a dose-dependent fashion against
bial, as one study found, where non-antimicrobial pro- carbapenem-resistant K. pneumoniae embedded in bio-
teins such as GFP and IFN-γ, when presented as IBs, films [20]. However, it is important to point out that the
achieved a significant reduction in bacterial loads [13]. selection of an optimal mild solubilizer is especially rel-
evant when antimicrobial proteins are purified because
IBs as a source of soluble HDPs recently it has been reported that they can impair the
IBs can also be used as an alternative source to obtain sol- antimicrobial activity [81].
uble HDPs when recombinant proteins aggregate (Fig. 3).
This is useful when the isolated soluble version of the Strategies to optimize HDP production
HDP is required for specific applications, but most of the Fusion tags
protein of interest forms aggregates. It is especially rel- There is a wide range of fusion partners (fusion tags) that
evant when HDPs are produced in E. coli, since in many have been used, as a strategy to properly express HDPs
cases it is necessary to recover the protein of interest in a soluble form (Table 1). These solubility enhanc-
from the aggregated fraction (Table 1). A high percentage ing domains assist with correct folding and promote the
(around 34%) of the recombinant HDPs produced in E. expression to the soluble fraction of the protein of inter-
coli are extracted from IBs (Table 1). est. In addition to enhance HDP solubility, tags might
In general terms, high concentrations of denatur- also protect against proteolysis, which HDPs are prone
ing agents such as 6M GdnHCl or 8M urea are used to to due to their small size [82]. Some widely used solu-
extract soluble protein from IBs. However, alternative bility tags that tend to yield high levels of HDPs in the
protocols have been developed in the last years [78]. cytoplasm of E. coli are GST, Trx, GFP, SUMO and Silk
Since it has been widely proven that proteins embedded (Table 1, Fig. 4A) [43, 83–88]. Although they need to be
in these nanoparticles can still be functional, the soluble studied empirically on a case to case basis, in general they
form of different proteins has also been extracted under considerably improve the solubility of many recombinant
mild, non-denaturing conditions [20, 66, 78–80]. Unlike proteins [89].

Fig. 3 Mild solubilization of HDP produced as IB in prokaryotic systems such as E. coli [20]

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


Roca‑Pinilla et al. Microbial Cell Factories (2022) 21:267 Page 9 of 15

Fig. 4 Different strategies allow the successful production of HDPs, which might otherwise be refractory to recombinant production. A A
fusion partner that mimics the peptide precursor structure, but that must be removed downstream, B self-cleavable tags such as inteins, C
specificity-targeting tags and D a multi-domain HDP combined or not with antimicrobial proteins and other non-antimicrobial domains

An interesting approach is the use of the N-terminal Multidomain HDPs


domain of spider silk as a fusion tag, which has been Some researchers have explored the use of fusion part-
proven to yield up to 8 times more soluble protein ners that add other functions that go beyond just helping
compared to other frequently used expression tags and to fold and express the recombinant HDPs. An example
allows the expression of otherwise difficult to express of this are cationic elastin-like polypeptides (ELP), that
peptides and proteins, such as the HDP precursor of have successfully been used to purify a fusion hybrid of
LL-37 (hCAP18) [90]. Alternatively, acidic peptides can cecropin A and D without the need of chromatography
also be used, since they offer a charge-charge interac- [97]. ELP tags allow to form reversible spherical aggre-
tion that neutralizes the potential bactericidal effect of gates that allow to precipitate the fused HDPs under cer-
HDPs, avoiding the death of the host microorganism tain temperature conditions, in a process called inverse
[91]. transition cycling, thus simplifying downstream process-
However, in many cases, there is a need to remove the ing, in addition to allowing HDP expression.
carrier protein to isolate the peptide of interest. And A different example, that shows how fusion partners
often, removing the fusion partner requires expensive might add new features to HDPs, is the broad-spectrum
enzymatic cleavage or toxic reagents, such as cyanogen plant defensin HDP C6, which can be fused to a peptide
bromide or enterokinase hydrolysis [92, 93]. pheromone (cCF10) to add specificity to the original
To overcome tag removal hurdles, self-cleaving tags HDP. The cCF10 pheromone domain is species-specific
(Fig. 4B), such as inteins can be used. Inteins are protein and binds to the bacterial membrane of Enterococcus
segments that can cut themselves from their precursors faecalis with high affinity, an appealing addition to the
and re-join the flanking regions, also known as protein original antimicrobial activity that allows for the precise
splicing [94]. This system has been used to express a killing of E. faecalis while avoiding potential off-target
cathelicidin, using B. subtilis as a host, allowing to purify killing of beneficial microorganisms found in the host
the HDP by affinity chromatography and self-cleave in microbiome, which is what most the conventional anti-
one step [95]. Self-cleaving tags, therefore, enable puri- biotics do [98]. This type of multidomain approach is also
fication and cleavage in a single step, saving time, labor known as specifically targeted antimicrobial peptides
and reducing cost, but have an inherent risk of incom- (STAMPs) [99]. Many combinations of killing and tar-
plete or uncontrolled cleavage [96]. geting domains can be tried, where wild-type, rationally

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


Roca‑Pinilla et al. Microbial Cell Factories (2022) 21:267 Page 10 of 15

enhanced or artificial sequences can be used [100–103]. individual components, namely catalytic, antimicrobial,
In all cases, these hybrid antimicrobials show improved and immune-modulatory activities [21, 104].
antimicrobial activity, selectivity, and kinetics against
their specific targets [87, 98]. Nonetheless, there still are Mutlidomain HDP fragment‑stitching
inevitable bactericidal effects on other bacteria. Like the multidomain approach, segments (but not the
The use of additional HDPs as fusion partners of full active sequence) of an HDP can be stitched together
other HDPs represents a unique strategy for the gen- in a hybrid molecule that is a mix of the parental pep-
eration of recombinant multidomain HDPs (Fig. 5D), tides, a process we named HDP fragment-stitching. Frag-
without the need for a carrier protein that needs to be ments of peptides such as the human cathelicidin LL-37,
removed downstream. For example, the pore-forming CM4 from a Chinese domestic silk moth and TP5, a frag-
HD5 has been fused to an enzyme that hydrolyses bac- ment of the thymus hormone, have been used in one
terial membrane phospholipids, such as human XII-A study to create hybrid antimicrobial that works against
secreted phospholipase A2 ­(sPLA2), generating a multi- enterotoxic E. coli [105]. Fragment-stitching might be
domain antimicrobial protein that can be successfully useful to remove undesired activities from a parental
expressed recombinantly, and that attacks bacteria by peptide, such as the hemolytic activity of LL-37, while
using two completely different mechanisms [20]. This generating new HDPs.
broad-spectrum multidomain construct, named JAMF1,
was effective against several antibiotic resistant bacterial Synergy of recombinant HDPs and antibiotics
strains such as quinolone and carbapenem resistant K. The performance of antibiotics against some antimicro-
pneumoniae (Fig. 5D). Similarly, in an effort to develop a bial resistant (AMR) bacteria or in bacterial living resist-
vaccine against glyceraldehyde-3-phosphate dehydroge- ant forms such as biofilms could be improved by their
nase (GAPDH), an enzyme involved in the virulence of combination with other drugs. Numerous studies have
Mycoplasma bovis, different bovine HDPs were fused to assessed this principle by using synthetic HDPs [106] and
GAPDH to boost the immune response against GAPDH antibiotics, demonstrating a clear synergy and beneficial
(i.e. BMAP28, TAP and indolicidin) [104]. Interest- effects on infection treatment. The main advantage of
ingly, when HDPs are linked to enzymes, such as in the this strategy is to reduce the dose of each drug and conse-
construct JAMF1 or GAPDH-HDP chimeras, both the quently the possible toxic effects and eventually combine
enzyme and the HDP seem to keep the activities of the different mechanisms to control bacterial survival along

Fig. 5 The boundless possibilities of multidomain HDPs. There is a vast range of domains to choose from for each domain we might want to add to
our chimeric HDP constructs, each one adding new functionalities, that might not necessarily be related to host defense (i.e. target specificity)

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


Roca‑Pinilla et al. Microbial Cell Factories (2022) 21:267 Page 11 of 15

with the emergence of bacterial resistances. In the con- Abbreviations


ARB: Antimicrobial resistant bacteria; AMP: Antimicrobial peptide; HDP: Host-
text of recombinant HDPs, not many studies have been defense peptide; SPPS: Solid-phase peptide synthesis; PTMS: Post-translational
carried out. However, if synthetic HDPs can synergize modifications; GRAS: Generally Recognized as Safe; CHO: Chinese Hamster
with antibiotics, so should their recombinant version. Ovary; HSA: Human serum albumin; AMR: Antimicrobial resistance; rMBD:
Recombinant mouse beta defensin; MRSA: Methicillin resistant S. aureus; IB:
To explore this, researchers tested the synergy of mouse Inclusion bodies; HD5: Human alpha defensin 5; CAP: Cysteine rich acidic pep‑
β-defensin 3 (rMBD3) with different antibiotics against tide; MIC: Minimal inhibitory concentration; PLGA: Poly(lactic-co-glycolic acid);
bacterial and yeast drug-resistant strains in vitro [107]. NAC: N-Acetyl cysteine; PEG: Polyethylene glycol; hCAP: Human cathelicidin
antimicrobial protein; ELP: Elastin-like polypeptides; cCF10: Cell-associated
Interestingly, they found that the anti-methicillin-resist- pheromone peptide; STAMPS: Specifically targeted antimicrobial peptides;
ant S. aureus (MRSA) activity of rMBD3 in combination sPLA2: Secreted phospholipase ­A2; GAPDH: Glyceraldehyde 3-phosphate
with ampicillin was synergistic, but it was not effective dehydrogenase.
against methicillin sensible S. aureus [107]. Combina- Acknowledgements
tions of rMBD3 with itraconazole, amphotericin or 5-flu- Figures were created with BioRender.com.
orocytosine were synergistic against two tested Candida
Author contributions
albicans strains. These results support the potential of RRP, AA and EGF conceived and designed the manuscript. RRP, AA and EGF
recombinant HDP to improve the activities of conven- performed the bibliographic research, conceptualized, and drafted the manu‑
tional antibiotics [107] and suggest that the same mecha- script. LL outlined the structure and reviewed the manuscript. All authors read
and approved the final manuscript.
nism that makes bacteria resistant to a certain antibiotic
might make them more vulnerable to combinatorial Funding
treatments. This work was funded by Ministerio de Ciencia, Innovación y Universidades
Grant (PID2019-107298RB-C21/AEI/10.13039/501100011033) to AA and EG-F
and by Marató de TV3 foundation (201812-30-31-32-33) to EG-F. The authors
Conclusions are also indebted to CERCA Programme (Generalitat de Catalunya) and Euro‑
pean Social Fund for supporting our research. RRP was supported by internal
HDPs hold promise as new candidates to combat anti- funding from Children’s Medical Research Institute.
biotic-resistant pathogens. There is a clear need for
new potent HDP candidates and the means to produce Availability of data and materials
The datasets supporting the review are referenced throughout the article.
them efficiently and at a low-cost. Research on different
expression systems will effectively accelerate our ability
Declarations
to increase production yields, peptide structure and bio-
activity and provide access to engineered microbial cell Ethics approval and consent to participate
factories that work universally well for most HDPs. But Not applicable.

the optimal expression systems are only one part of the Consent for publication
equation. Not applicable.
New recombinant HDP forms, such as IBs and encap-
Competing interests
sulated HDPs, are also worth considering. They offer The authors have no competing interests to disclose.
properties that the soluble form does not provide, includ-
ing higher stability, a slower release profile, reduced HDP Author details
1
Translational Vectorology Research Unit, Faculty of Medicine and Health, Chil‑
toxicity or on-site activation. Besides, IBs should be dren’s Medical Research Institute, The University of Sydney, Westmead, NSW
exploited as both a new antimicrobial HDP format and 2145, Australia. 2 Laboratory of Molecular Oncology and Innovative Therapies,
as a treasure trove of bioactive, functional, and soluble Military Institute of Medicine, Warsaw, Poland. 3 Department of Ruminant Pro‑
duction, Institut de Recerca i Tecnologia Agroalimentàries IRTA​, 08140 Caldes
HDPs. de Montbui, Spain.
Finally, strategies to optimize HDP production, such
as fusion tags and multidomain HDPs, open the door to Received: 7 November 2022 Accepted: 10 December 2022

designing newly added functionalities, such as the pre-


cise killing of a pathogen, while avoiding off-target effects
on probiotic bacteria. Moreover, a well-crafted tag strat-
References
egy not only increases the uses of a recombinant HDP 1. Antimicrobial Resistance Collaborators. Global burden of bacte‑
but also should allow for its production without the need rial antimicrobial resistance in 2019: a systematic analysis. Lancet.
of downstream tag removal. Collectively, finely-tuned 2022;399:629–55.
2. Lázár V, Martins A, Spohn R, Daruka L, Grézal G, Fekete G, et al.
recombinant approaches show promise for much-needed Antibiotic-resistant bacteria show widespread collateral sensitivity to
sustainable, inexpensive, and larger-scale production of antimicrobial peptides. Nat Microbiol. 2018;3:718–31.
antimicrobial peptides as well as other peptide therapeu- 3. Zasloff M. Antimicrobial peptides of multicellular organisms. Nature.
2002;415:389–95.
tics and in turn, allow these molecules to enter clinical 4. Hancock REW, Sahl HG. Antimicrobial and host-defense pep‑
practice. tides as new anti-infective therapeutic strategies. Nat Biotechnol.
2006;24:1551–7.

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


Roca‑Pinilla et al. Microbial Cell Factories (2022) 21:267 Page 12 of 15

5. Steiner H, Hultmark D, Engström A, Bennich H, Boman HG. Sequence 27. Haney EF, Straus SK, Hancock REW. Reassessing the host defense pep‑
and specificity of two antibacterial proteins involved in insect immu‑ tide landscape. Front Chem. 2019;7:43.
nity. Nature. 1981;292:246–8. 28. Drayton M, Kizhakkedathu JN, Straus SK. Towards robust delivery of
6. Lehrer RI, Szklarek D, Ganz T, Selsted ME. Correlation of binding of rabbit antimicrobial peptides to combat bacterial resistance. Molecules.
granulocyte peptides to Candida albicans with candidacidal activity. 2020;25:3048.
Infect Immun. 1985;49:207–11. 29. Münzker L, Oddo A, Hansen PR. Chemical synthesis of antimicro‑
7. Zasloff M. Antimicrobial peptides of multicellular organisms: my per‑ bial peptides. Methods Mol Biol. 2017. https://​doi.​org/​10.​1007/​
spective. Adv Exp Med Biol. 2019;1117:3–6. 978-1-​4939-​6737-7_3.
8. Wang G, Li X, Wang Z. APD3: the antimicrobial peptide database as a 30. da Costa JP, Cova M, Ferreira R, Vitorino R. Antimicrobial peptides:
tool for research and education. Nucleic Acids Res. 2016;44:D1087–93. an alternative for innovative medicines? Appl Microbiol Biotechnol.
9. Torrent M, Valle J, Nogués MV, Boix E, Andreu D. The generation of anti‑ 2015;99:2023–40.
microbial peptide activity: a trade-off between charge and aggrega‑ 31. Wibowo D, Zhao C-X. Recent achievements and perspectives for
tion? Angew Chem Int Ed Engl. 2011;50:10686–9. large-scale recombinant production of antimicrobial peptides. Appl
10. Mookherjee N, Anderson MA, Haagsman HP, Davidson DJ. Antimicrobial Microbiol Biotechnol. 2019;103:659–71.
host defence peptides: functions and clinical potential. Nat Rev Drug 32. Latham PW. Therapeutic peptides revisited. Nat Biotechnol.
Discov. 2020;19:311–32. 1999;17:755–7.
11. Cândido ES, Cardoso MH, Chan LY, Torres MDT, Oshiro KGN, Porto 33. Sampaio de Oliveira KB, Leite ML, Rodrigues GR, Duque HM, da
WF, et al. Short cationic peptide derived from archaea with dual Costa RA, Cunha VA, et al. Strategies for recombinant production of
antibacterial properties and anti-infective potential. ACS Infect Dis. antimicrobial peptides with pharmacological potential. Expert Rev Clin
2019;5:1081–6. Pharmacol. 2020;13:367–90.
12. Dassanayake RP, Falkenberg SM, Register KB, Samorodnitsky D, Nichol‑ 34. Kaur J, Kumar A, Kaur J. Strategies for optimization of heterologous
son EM, Reinhardt TA. Antimicrobial activity of bovine NK-lysin-derived protein expression in E. coli: roadblocks and reinforcements. Int J Biol
peptides on Mycoplasma bovis. PLoS ONE. 2018;13:1–17. Macromol. 2018;106:803–22.
13. Carratalá JV, Brouillette E, Serna N, Sánchez-Chardi A, Sánchez JM, 35. Parachin NS, Mulder KC, Viana AAB, Dias SC, Franco OL. Expression sys‑
Villaverde A, et al. In vivo bactericidal efficacy of GWH1 antimicrobial tems for heterologous production of antimicrobial peptides. Peptides.
peptide displayed on protein nanoparticles, a potential alternative to 2012;38:446–56.
antibiotics. Pharmaceutics. 2020;12:1217. 36. Deng T, Ge H, He H, Liu Y, Zhai C, Feng L, et al. The heterologous expres‑
14. Mishra B, Wang G. Individual and combined effects of engineered pep‑ sion strategies of antimicrobial peptides in microbial systems. Protein
tides and antibiotics on Pseudomonas aeruginosa biofilms. Pharmaceu‑ Expr Purif. 2017;140:52–9.
ticals. 2017;10:58. 37. López-Cano A, Martínez-Miguel M, Guasch J, Ratera I, Arís A, Garcia-
15. De La Fuente-Núñez C, Cardoso MH, De Souza CE, Franco OL, Hancock Fruitós E. Exploring the impact of the recombinant Escherichia coli strain
REW. Synthetic antibiofilm peptides. Biochim Biophys Acta Biomembr. on defensins antimicrobial activity: BL21 versus Origami strain. Microb
2016;1858:1061–9. Cell Fact. 2022;21:77.
16. Freitas CG, Lima SMF, Freire MS, Cantuária APC, Júnior NGO, Santos TS, 38. Sørensen HP, Mortensen KK. Advanced genetic strategies for
et al. An immunomodulatory peptide confers protection in an experi‑ recombinant protein expression in Escherichia coli. J Biotechnol.
mental candidemia murine model. Antimicrob Agents Chemother. 2005;115:113–28.
2017;61: e02518-16. 39. Deo S, Turton KL, Kainth T, Kumar A, Wieden H-J. Strategies for improv‑
17. van Harten R, van Woudenbergh E, van Dijk A, Haagsman H. Cathelici‑ ing antimicrobial peptide production. Biotechnol Adv. 2022;59: 107968.
dins: immunomodulatory antimicrobials. Vaccines. 2018;6:63. 40. Kuddus MDR, Rumi F, Tsutsumi M, Takahashi R, Yamano M, Kamiya M,
18. Hancock REW, Haney EF, Gill EE. The immunology of host defence pep‑ et al. Expression, purification and characterization of the recombinant
tides: beyond antimicrobial activity. Nat Rev Immunol. 2016;16:321–34. cysteine-rich antimicrobial peptide snakin-1 in Pichia pastoris. Protein
19. Sandín D, Valle J, Chaves-Arquero B, Prats-Ejarque G, Larrosa MN, Expr Purif. 2016;122:15–22.
González-López JJ, et al. Rationally modified antimicrobial peptides 41. Bommarius B, Jenssen H, Elliott M, Kindrachuk J, Pasupuleti M, Gieren
from the N-terminal domain of human RNase 3 show exceptional H, et al. Cost-effective expression and purification of antimicrobial and
serum stability. J Med Chem. 2021;64:11472–82. host defense peptides in Escherichia coli. Peptides. 2010;31:1957–65.
20. Roca-Pinilla R, López-Cano A, Saubi C, Garcia-Fruitós E, Arís A. A 42. Yong E. Yeast suggests speedy start for multicellular life. Nature. 2012.
new generation of recombinant polypeptides combines multiple https://​doi.​org/​10.​1038/​nature.​2012.​9810.
protein domains for effective antimicrobial activity. Microb Cell Fact. 43. Cao J, de la Fuente-Nunez C, Ou RW, Torres MDT, Pande SG, Sinskey AJ,
2020;19:1–7. et al. Yeast-based synthetic biology platform for antimicrobial peptide
21. Roca-Pinilla R, Holani R, López-Cano A, Saubi C, Baltà-Foix R, Cobo ER, production. ACS Synth Biol. 2018;7:896–902.
et al. Sequence edition of single domains modulates the final immune 44. Tai H-M, You M-F, Lin C-H, Tsai T-Y, Pan C-Y, Chen J-Y. Scale-up produc‑
and antimicrobial potential of a new generation of multidomain tion of and dietary supplementation with the recombinant antimicro‑
recombinant proteins. Sci Rep. 2021;11:23798. bial peptide tilapia piscidin 4 to improve growth performance in Gallus
22. Silva ON, Torres MDT, Cao J, Alves ESF, Rodrigues LV, Resende JM, et al. gallus domesticus. PLoS ONE. 2021;16: e0253661.
Repurposing a peptide toxin from wasp venom into antiinfectives with 45. Zhang X, Jiang A, Qi B, Yu H, Xiong Y, Zhou G, et al. Secretion expres‑
dual antimicrobial and immunomodulatory properties. Proc Natl Acad sion of human neutrophil peptide 1 (HNP1) in Pichia pastoris and its
Sci USA. 2020;117:26936–45. functional analysis against antibiotic-resistant Helicobacter pylori. Appl
23. De Santis E, Alkassem H, Lamarre B, Faruqui N, Bella A, Noble JE, et al. Microbiol Biotechnol. 2018;102:4817–27.
Antimicrobial peptide capsids of de novo design. Nat Commun. 46. Liang X, Jiang H, Si X, Xin Q, Meng D, Chen P, et al. Boosting expression
2017;8:2263. level of plectasin in recombinant Pichia pastoris via 2A self-processing
24. Torres MDT, Melo MCR, Crescenzi O, Notomista E, de la Fuente-Nunez C. peptide assembly. Appl Microbiol Biotechnol. 2022;106(9–10):3669–78.
Mining for encrypted peptide antibiotics in the human proteome. Nat 47. Thyab Gddoa Al-sahlany S, Altemimi A, Al-Manhel A, Niamah A,
Biomed Eng. 2022;6:67–75. Lakhssassi N, Ibrahim S. Purification of bioactive peptide with anti‑
25. Gudmundsson GH, Agerberth B, Odeberg J, Bergman T, Olsson B, microbial properties produced by Saccharomyces cerevisiae. Foods.
Salcedo R. The human gene FALL39 and processing of the cathelin pre‑ 2020;9:324.
cursor to the antibacterial peptide LL-37 in granulocytes. Eur J Biochem. 48. Xia L, Liu Z, Ma J, Sun S, Yang J, Zhang F. Expression, purification and
1996;238:325–32. characterization of cecropin antibacterial peptide from Bombyx mori in
26. Cesaro A, Torres MDT, Gaglione R, Dell’Olmo E, Di Girolamo R, Bosso Saccharomyces cerevisiae. Protein Expr Purif. 2013;90:47–54.
A, et al. Synthetic antibiotic derived from sequences encrypted in a 49. Mygind PH, Fischer RL, Schnorr KM, Hansen MT, Sönksen CP, Ludvigsen
protein from human plasma. ACS Nano. 2022;16(2):1880–95. S, et al. Plectasin is a peptide antibiotic with therapeutic potential from
a saprophytic fungus. Nature. 2005;437:975–80.

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


Roca‑Pinilla et al. Microbial Cell Factories (2022) 21:267 Page 13 of 15

50. Zhang M, Shan Y, Gao H, Wang B, Liu X, Dong Y, et al. Expression of a elucidating their inflammatory effects in vivo and their potential as a
recombinant hybrid antimicrobial peptide magainin II-cecropin B in the new therapeutic format. Pharmaceutics. 2020;12:450.
mycelium of the medicinal fungus Cordyceps militaris and its validation 70. Villaverde A, Corchero JL, Seras-Fanzoso J, Garcia-Fruitós E. Func‑
in mice. Microb Cell Fact. 2018;17:18. tional protein aggregates: just the tip of the iceberg. Nanomedicine.
51. Carballar-Lejarazú R, Rodríguez MH, de la Cruz H-H, Ramos-Castañeda J, 2015;10:2881–91.
Possani LD, Zurita-Ortega M, et al. Recombinant scorpine: a multifunc‑ 71. Rueda F, Gasser B, Sánchez-Chardi A, Roldán M, Villegas S, Puxbaum V,
tional antimicrobial peptide with activity against different pathogens. et al. Functional inclusion bodies produced in the yeast Pichia pastoris.
Cell Mol Life Sci. 2008;65:3081–92. Microb Cell Fact. 2016;15:166.
52. Zitzmann J, Weidner T, Czermak P. Optimized expression of the 72. Seras-Franzoso J, Cano-Garrido O, Peternel S, Arís A, Garcia-Fruitós
antimicrobial protein Gloverin from Galleria mellonella using stably E. Purification of inclusion bodies produced in bacteria and yeast. Meth‑
transformed Drosophila melanogaster S2 cells. Cytotechnology. ods Mol Biol. 2022;2406:401–16.
2017;69:371–89. 73. Pesarrodona M, Jauset T, Díaz-Riascos ZV, Sánchez-Chardi A, Beaulieu
53. Hu Y. Baculovirus as a highly efficient expression vector in insect and M, Seras-Franzoso J, et al. Targeting antitumoral proteins to breast
mammalian cells. Acta Pharmacol Sin. 2005;26:405–16. cancer by local administration of functional inclusion bodies. Adv Sci.
54. Ingham AB, Moore RJ. Recombinant production of antimicrobial 2019;6:1900849.
peptides in heterologous microbial systems. Biotechnol Appl Biochem. 74. Hrabárová E, Achbergerová L, Nahálka J. Insoluble protein applications:
2007;47:1–9. the use of bacterial inclusion bodies as biocatalysts. New York: Humana
55. Li Y, Chen Z. RAPD: a database of recombinantly-produced antimicro‑ Press; 2015. p. 411–22. https://​doi.​org/​10.​1007/​978-1-​4939-​2205-5_​24.
bial peptides. FEMS Microbiol Lett. 2008;289:126–9. 75. Seras-Franzoso J, Peebo K, García-Fruitós E, Vázquez E, Rinas U, Vil‑
56. Rinas U, Garcia-Fruitós E, Corchero JL, Vázquez E, Seras-Franzoso J, laverde A. Improving protein delivery of fibroblast growth factor-2 from
Villaverde A. Bacterial inclusion bodies: discovering their better half. bacterial inclusion bodies used as cell culture substrates. Acta Biomater.
Trends Biochem Sci. 2017;42:726–37. 2014;10:1354–9.
57. de Marco A, Ferrer-Miralles N, Garcia-Fruitós E, Mitraki A, Peternel S, 76. Torrealba D, Parra D, Seras-Franzoso J, Vallejos-Vidal E, Yero D, Gibert I,
Rinas U, et al. Bacterial inclusion bodies are industrially exploitable et al. Nanostructured recombinant cytokines: a highly stable alternative
amyloids. FEMS Microbiol Rev. 2019;43:53–72. to short-lived prophylactics. Biomaterials. 2016;107:102–14.
58. van Gent ME, Ali M, Nibbering PH, Kłodzińska SN. Current advances 77. Gifre-Renom L, Seras-Franzoso J, Rafael D, Andrade F, Cano-Garrido O,
in lipid and polymeric antimicrobial peptide delivery systems and Martinez-Trucharte F, et al. The biological potential hidden in inclusion
coatings for the prevention and treatment of bacterial infections. bodies. Pharmaceutics. 2020;12:157.
Pharmaceutics. 2021;13:1840. 78. Singhvi P, Saneja A, Srichandan S, Panda AK. Bacterial inclusion bodies: a
59. Sharma A, Vaghasiya K, Ray E, Verma RK. Nano-encapsulated HHC10 treasure trove of bioactive proteins. Trends Biotechnol. 2020;38:474–86.
host defense peptide (HDP) reduces the growth of Escherichia 79. Gifre-Renom L, Cano-Garrido O, Fàbregas F, Roca-Pinilla R, Seras-
coli via multimodal mechanisms. Artif Cells Nanomed Biotechnol. Franzoso J, Ferrer-Miralles N, et al. A new approach to obtain pure and
2018;46:S156–65. active proteins from Lactococcus lactis protein aggregates. Sci Rep.
60. Sharma A, Vaghasiya K, Ray E, Gupta P, Kumar Singh A, Datta Gupta U, 2018;8:1–10.
et al. Mycobactericidal activity of some micro-encapsulated synthetic 80. Singhvi P, Saneja A, Ahuja R, Panda AK. Solubilization and refolding of
host defense peptides (HDP) by expediting the permeation of anti‑ variety of inclusion body proteins using a novel formulation. Int J Biol
biotic: a new paradigm of drug delivery for tuberculosis. Int J Pharm. Macromol. 2021;193:2352–64.
2019;558:231–41. 81. López-Cano A, Sicilia P, Gaja C, Arís A, Garcia-Fruitós E. Quality com‑
61. Sharma A, Vaghasiya K, Gupta P, Singh AK, Gupta UD, Verma RK. parison of recombinant soluble proteins and proteins solubilized from
Dynamic mucus penetrating microspheres for efficient pulmonary bacterial inclusion bodies. N Biotechnol. 2022;72:58–63.
delivery and enhanced efficacy of host defence peptide (HDP) in 82. Li Y. Carrier proteins for fusion expression of antimicrobial peptides in
experimental tuberculosis. J Control Release. 2020;324:17–33. Escherichia coli. Biotechnol Appl Biochem. 2009;54:1–9.
62. Javia A, Misra A, Thakkar H. Liposomes encapsulating novel antimicro‑ 83. Baker RT. Protein expression using ubiquitin fusion and cleavage. Curr
bial peptide Omiganan: characterization and its pharmacodynamic Opin Biotechnol. 1996;7:541–6.
evaluation in atopic dermatitis and psoriasis mice model. Int J Pharm. 84. Hammarström M, Hellgren N, van den Berg S, Berglund H, Härd T. Rapid
2022;624: 122045. screening for improved solubility of small human proteins produced as
63. Reijmar K, Edwards K, Andersson K, Agmo HV. Characterizing and con‑ fusion proteins in Escherichia coli. Protein Sci. 2009;11:313–21.
trolling the loading and release of cationic amphiphilic peptides onto 85. Dyson MR, Shadbolt SP, Vincent KJ, Perera RL, McCafferty J. Production
and from PEG-stabilized lipodisks. Langmuir. 2016;32:12091–9. of soluble mammalian proteins in Escherichia coli: identification of pro‑
64. Kaur N, Dilawari R, Kaur A, Sahni G, Rishi P. Recombinant expres‑ tein features that correlate with successful expression. BMC Biotechnol.
sion, purification and PEGylation of Paneth cell peptide (cryptdin-2) 2004;4:32.
with value added attributes against Staphylococcus aureus. Sci Rep. 86. Satakarni M, Curtis R. Production of recombinant peptides as fusions
2020;10:12164. with SUMO. Protein Expr Purif. 2011;78:113–9.
65. Drayton M, Alford MA, Pletzer D, Haney EF, Machado Y, Luo HD, et al. 87. Wang M, Zheng K, Lin J, Huang M, Ma Y, Li S, et al. Rapid and efficient
Enzymatically releasable polyethylene glycol—host defense peptide production of cecropin A antibacterial peptide in Escherichia coli by
conjugates with improved activity and biocompatibility. J Control fusion with a self-aggregating protein. BMC Biotechnol. 2018;18:62.
Release. 2021;339:220–31. 88. Sinha R, Shukla P. Antimicrobial peptides: recent insights on biotech‑
66. Gifre-Renom L, Baltà-Foix R, Arís A, Garcia-Fruitós E. Nondenaturing nological interventions and future perspectives. Protein Pept Lett.
solubilization of inclusion bodies from lactic acid bacteria. In: Insoluble 2019;26:79–87.
proteins 2022. New York: Humana; 2022. p. 389–400. 89. van Ham TJ, Esposito A, Kumita JR, Hsu STD, Kaminski Schierle GS,
67. López-Cano A, Bach A, López-Serrano S, Aragon V, Blanch M, Pas‑ Kaminski CF, et al. Towards multiparametric fluorescent imaging of
tor JJ, et al. Potential of oral nanoparticles containing cytokines as amyloid formation: studies of a YFP model of α-synuclein aggregation. J
intestinal mucosal immunostimulants in pigs: a pilot study. Animals. Mol Biol. 2010;395:627–42.
2022;12:1075. 90. Kronqvist N, Sarr M, Lindqvist A, Nordling K, Otikovs M, Venturi L, et al.
68. Cano-Garrido O, Sánchez-Chardi A, Parés S, Giró I, Tatkiewicz WI, Ferrer- Efficient protein production inspired by how spiders make silk. Nat
Miralles N, et al. Functional protein-based nanomaterial produced in Commun. 2017;8:15504.
microorganisms recognized as safe: a new platform for biotechnology. 91. Kim SC, Jang SA, Sung BH, Kim JM, Lim KJ, Shin JR, et al. Method for the
Acta Biomater. 2016;43:230–9. mass expression of an antimicrobial peptide by co-expression of a basic
69. Gifre-Renom L, Ugarte-Berzal E, Martens E, Boon L, Cano-Garrido O, antimicrobial peptide and an acidic peptide using a translational cou‑
Martínez-Núñez E, et al. Recombinant protein-based nanoparticles: pling system. US: Korea Advanced Inst Sci & Tech OP—KR 20070141932.

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


Roca‑Pinilla et al. Microbial Cell Factories (2022) 21:267 Page 14 of 15

92. Lee JH, Minn I, Park CB, Kim SC. Acidic peptide-mediated expression of 114. Popa C, Shi X, Ruiz T, Ferrer P, Coca M. Biotechnological production of
the antimicrobial peptide buforin II as tandem repeats in Escherichia the cell penetrating antifungal PAF102 peptide in Pichia pastoris. Front
coli. Protein Expr Purif. 1998;12:53–60. Microbiol. 2019;10:1472.
93. Srinivasulu B, Syvitski R, Seo JK, Mattatall NR, Knickle LC, Douglas SE. 115. Panteleev PV, Bolosov IA, Kalashnikov AÀ, Kokryakov VN, Shamova OV,
Expression, purification and structural characterization of recombinant Emelianova AA, et al. combined antibacterial effects of goat cathelici‑
hepcidin, an antimicrobial peptide identified in Japanese flounder, dins with different mechanisms of action. Front Microbiol. 2018;9:2983.
Paralichthys olivaceus. Protein Expr Purif. 2008;61:36–44. 116. Al Kashgry NAT, Abulreesh HH, El-Sheikh IA, Almaroai YA, Salem R,
94. Perler FB, Davis EO, Dean GE, Gimble FS, Jack WE, Neff N, et al. Protein Mohamed I, et al. Utilization of a recombinant defensin from Maize (Zea
splicing elements: inteins and exteins—a definition of terms and mays L.) as a potential antimicrobial peptide. AMB Express. 2020;10:208.
recommended nomenclature. Nucleic Acids Res. 1994;22:1125–7. 117. Li Z-J, Zhang Z-X, Xu Y, Shi T-Q, Ye C, Sun X-M, et al. CRISPR-based
95. He Q, Fu A, Li T. Expression and one-step purification of the antimicro‑ construction of a BL21 (DE3)-derived variant strain library to
bial peptide cathelicidin-BF using the intein system in Bacillus subtilis. J rapidly improve recombinant protein production. ACS Synth Biol.
Ind Microbiol Biotechnol. 2015;42:647–53. 2022;11:343–52.
96. Li Y. Self-cleaving fusion tags for recombinant protein production. 118. Hao X, Chi H, Tang X, Xing J, Sheng X, Zhan W. The functions of
Biotechnol Lett. 2011;33:869–81. β-defensin in flounder (Paralichthys olivaceus): antibiosis, chemotaxis
97. Yang K, Su Y, Li J, Sun J, Yang Y. Expression and purification of the and modulation of phagocytosis. Biology. 2021;10:1247.
antimicrobial peptide cecropin AD by fusion with cationic elastin-like 119. Ishida H, Nguyen LT, Gopal R, Aizawa T, Vogel HJ. Overexpression of
polypeptides. Protein Expr Purif. 2012;85:200–3. antimicrobial, anticancer, and transmembrane peptides in Escherichia
98. Xu L, Shao C, Li G, Shan A, Chou S, Wang J, et al. Conversion of broad- coli through a calmodulin-peptide fusion system. J Am Chem Soc.
spectrum antimicrobial peptides into species-specific antimicrobi‑ 2016;138:11318–26.
als capable of precisely targeting pathogenic bacteria. Sci Rep. 120. Yang N, Teng D, Mao R, Hao Y, Wang X, Wang Z, et al. A recombi‑
2020;10:944. nant fungal defensin-like peptide-P2 combats multidrug-resistant
99. Sarma P, Mahendiratta S, Prakash A, Medhi B. Specifically targeted Staphylococcus aureus and biofilms. Appl Microbiol Biotechnol.
antimicrobial peptides: a new and promising avenue in selective anti‑ 2019;103:5193–213.
microbial therapy. Indian J Pharmacol. 2018;50:1. 121. Acosta S, Quintanilla L, Alonso M, Aparicio C, Rodríguez-Cabello JC.
100. Xi D, Teng D, Wang X, Mao R, Yang Y, Xiang W, et al. Design, expression Recombinant AMP/polypeptide self-assembled monolayers with syner‑
and characterization of the hybrid antimicrobial peptide LHP7, con‑ gistic antimicrobial properties for bacterial strains of medical relevance.
nected by a flexible linker, against Staphylococcus and Streptococcus. ACS Biomater Sci Eng. 2019;5:4708–16.
Process Biochem. 2013;48:453–61. 122. Xie Q, Wang Y, Zhang M, Wu S, Wei W, Xiao W, et al. Recombinant HNP-1
101. He J, Yarbrough DK, Kreth J, Anderson MH, Shi W, Eckert R. Systematic produced by Escherichia coli triggers bacterial apoptosis and exhibits
approach to optimizing specifically targeted antimicrobial pep‑ antibacterial activity against drug-resistant bacteria. Microbiol Spectr.
tides against Streptococcus mutans. Antimicrob Agents Chemother. 2022;10: e00860-21.
2010;54:2143–51. 123. Wang A, Wang S, Shen M, Chen F, Zou Z, Ran X, et al. High level expres‑
102. He J, Anderson MH, Shi W, Eckert R. Design and activity of a ‘dual- sion and purification of bioactive human α-defensin 5 mature peptide
targeted’ antimicrobial peptide. Int J Antimicrob Agents. 2009;33:532–7. in Pichia pastoris. Appl Microbiol Biotechnol. 2009;84:877–84.
103. Eckert R, Qi F, Yarbrough DK, He J, Anderson MH, Shi W. Adding 124. Čipáková I, Hostinová E, Gašperı́k J, Velebný V. High-level expression and
selectivity to antimicrobial peptides: rational design of a multidomain purification of a recombinant hBD-1 fused to LMM protein in Escheri-
peptide against Pseudomonas spp. Antimicrob Agents Chemother. chia coli protein. Expr Purif. 2004;37:207–12.
2006;50:1480–8. 125. Maiti S, Patro S, Purohit S, Jain S, Senapati S, Dey N. Effective control
104. van der Merwe J, Prysliak T, Gerdts V, Perez-Casal J. Protein chimeras of Salmonella infections by employing combinations of recombinant
containing the Mycoplasma bovis GAPDH protein and bovine host- antimicrobial human β-defensins hBD-1 and hBD-2. Antimicrob Agents
defence peptides retain the properties of the individual components. Chemother. 2014;58:6896–903.
Microb Pathog. 2011;50:269–77. 126. Cipakova I, Hostinova E. Production of the human-beta-defensin using
105. Cheng J, Ahmat M, Guo H, Wei X, Zhang L, Cheng Q, et al. Expression, Saccharomyces cerevisiae as a host. Protein Pept Lett. 2005;12:551–4.
purification and characterization of a novel hybrid peptide CLP with 127. Lin Q, Xie K, Chen D, Yu B, Mao X, Yu J, et al. Expression and functional
excellent antibacterial activity. Molecules. 2021;26:7142. characterization of a novel antimicrobial peptide: human beta-defensin
106. Li J, Fernández-Millán P, Boix E. Synergism between host defence pep‑ 118. Biomed Res Int. 2020. https://​doi.​org/​10.​1155/​2020/​13953​04.
tides and antibiotics against bacterial infections. Curr Top Med Chem. 128. Xu Z, Peng L, Zhong Z, Fang X, Cen P. High-level expression of a soluble
2020;20:1238–63. functional antimicrobial peptide, human β-defensin 2, in Escherichia
107. Jiang Y, Yi X, Li M, Wang T, Qi T, She X. Antimicrobial activities of recom‑ coli. Biotechnol Prog. 2006;22:382–6.
binant mouse β-defensin 3 and its synergy with antibiotics. J Mater Sci 129. Peng L, Xu Z, Fang X, Wang F, Yang S, Cen P. Preferential codons enhanc‑
Mater Med. 2012;23:1723–8. ing the expression level of human beta-defensin-2 in recombinant
108. Behrendt R, White P, Offer J. Advances in Fmoc solid-phase peptide Escherichia coli. Protein Pept Lett. 2004;11:339–44.
synthesis. J Pept Sci. 2016;22:4–27. 130. Vargues T, Morrison G, Seo E, Clarke D, Fielder H, Bennani J, et al.
109. Yi T, Sun S, Huang Y, Chen Y. Prokaryotic expression and mechanism of Efficient production of human β-defensin 2 (HBD2) in Escherichia coli.
action of aα-helical antimicrobial peptide A20L using fusion tags. BMC Protein Pept Lett. 2009;16:668–76.
Biotechnol. 2015;15:1–10. 131. Wang F, Fang X, Xu Z, Peng L, Cen P. Fusion expression of human beta-
110. Meng D-M, Zhao J-F, Ling X, Dai H-X, Guo Y-J, Gao X-F, et al. Recom‑ defensin-2 from multiple joined genes in Escherichia coli. Prep Biochem
binant expression, purification and antimicrobial activity of a novel Biotechnol. 2004;34:215–25.
antimicrobial peptide PaDef in Pichia pastoris. Protein Expr Purif. 132. Huang L, Wang J, Zhong Z, Peng L, Chen H, Xu Z, et al. Production
2017;130:90–9. of bioactive human β-defensin-3 in Escherichia coli by soluble fusion
111. Zhou X, Wang Y, Pan Y, Li W. Nisin-controlled extracellular produc‑ expression. Biotechnol Lett. 2006;28:627–32.
tion of apidaecin in Lactococcus lactis. Appl Microbiol Biotechnol. 133. Xu Z, Zhong Z, Huang L, Peng L, Wang F, Cen P. High-level production
2008;78:947–53. of bioactive human beta-defensin-4 in Escherichia coli by soluble fusion
112. Stambuk F, Ojeda C, Machado Matos G, Rosa RD, Mercado L, Schmitt P. expression. Appl Microbiol Biotechnol. 2006;72:471–9.
Big defensin from the scallop Argopecten purpuratus ApBD1 is an anti‑ 134. Wang A, Su Y, Wang S, Shen M, Chen F, Chen M, et al. High efficiency
microbial peptide which entraps bacteria through nanonets formation. preparation of bioactive human α-defensin 6 in Escherichia coli
Fish Shellfish Immunol. 2021;119:456–61. Origami(DE3)pLysS by soluble fusion expression. Appl Microbiol Bio‑
113. Gong T, Du J, Li S-W, Huang H, Qi X-L. Identification and functional technol. 2010;87:1935–42.
analysis of a defensin CcDef2 from Coridius chinensis. Int J Mol Sci.
2022;23:2789.

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


Roca‑Pinilla et al. Microbial Cell Factories (2022) 21:267 Page 15 of 15

135. Huang L, Ching CB, Jiang R, Leong SSJ. Production of bioactive human 156. Tanhaeian A, Azghandi M, Mousavi Z, Javadmanesh A. Expression of
beta-defensin 5 and 6 in Escherichia coli by soluble fusion expression. thanatin in HEK293 cells and investigation of its antibacterial effects on
Protein Expr Purif. 2008;61:168–74. some human pathogens. Protein Pept Lett. 2019;27:41–7.
136. Liu H, Diao H, Hou J, Yu H, Wen H. Soluble expression and purification of 157. Luan C, Zhang HW, Song DG, Xie YG, Feng J, Wang YZ. Expressing
human β-defensin DEFB136 in Escherichia coli and identification of its antimicrobial peptide cathelicidin-BF in Bacillus subtilis using SUMO
bioactivity. Protein Expr Purif. 2021;188: 105968. technology. Appl Microbiol Biotechnol. 2014;98:3651–8.
137. Dai J, Ou W, Yu G, Ai Q, Zhang W, Mai K, et al. The antimicrobial peptide
cecropin AD supplement alleviated soybean meal-induced intestinal
inflammation, barrier damage, and microbial dysbiosis in juvenile Publisher’s Note
turbot, Scophthalmus maximus. Front Mar Sci. 2020;7: 584482. Springer Nature remains neutral with regard to jurisdictional claims in pub‑
138. Reichhart J-M, Petit I, Legrain M, Dimarcq J-L, Keppi E, Lecocq J-P, et al. lished maps and institutional affiliations.
Expression and secretion in yeast of active insect defensin, an inducible
antibacterial peptide from the fleshfly Phormia terranovae. Invertebr
Reprod Dev. 1992;21:15–24.
139. Perez-Perez DA, Villanueva-Ramirez TDJ, Hernandez-Pedraza AE, Casil‑
las-Vega NG, Gonzalez-Barranco P, Zarate X. The small metal-binding
protein SmbP simplifies the recombinant expression and purification of
the antimicrobial peptide LL-37. Antibiotics. 2021;10:1271.
140. Lin C-H, Pan Y-C, Liu F-W, Chen C-Y. Prokaryotic expression and action
mechanism of antimicrobial LsGRP1C recombinant protein containing
a fusion partner of small ubiquitin-like modifier. Appl Microbiol Biotech‑
nol. 2017;101:8129–38.
141. Hu J, Li S, Lv Q, Miao M, Li X, Li F. Characterization of the dual functions
of LvCrustinVII from Litopenaeus vannamei as antimicrobial peptide and
opsonin. Mar Drugs. 2022;20:157.
142. Li J, Islam S, Guo P, Hu X, Dong W. Isolation of antimicrobial genes from
Oryza rufipogon Griff by using a Bacillus subtilis expression system with
potential antimicrobial activities. Int J Mol Sci. 2020;21:8722.
143. Xu J, Zhong F, Zhang Y, Zhang J, Huo S, Lin H, et al. Construc‑
tion of Bacillus subtilis strain engineered for expression of porcine
β-defensin-2/cecropin P1 fusion antimicrobial peptides and its growth-
promoting effect and antimicrobial activity. Asian-Australas J Anim Sci.
2016;30:576–84.
144. Haught C, Davis GD, Subramanian R, Jackson KW, Harrison RG. Recom‑
binant production and purification of novel antisense antimicrobial
peptide in Escherichia coli. Biotechnol Bioeng. 1998;57:55–61.
145. Sun B, Wibowo D, Sainsbury F, Zhao C-X. Design and production of a
novel antimicrobial fusion protein in Escherichia coli. Appl Microbiol
Biotechnol. 2018;102:8763–72.
146. Trueman HE, Sriskantha A, Qu Y, Rapson TD, Sutherland TD. Modification
of honeybee silk by the addition of antimicrobial agents. ACS Omega.
2017;2:4456–63.
147. Zhang L, Li X, Wei D, Wang J, Shan A, Li Z. Expression of plectasin in
Bacillus subtilis using SUMO technology by a maltose-inducible vector. J
Ind Microbiol Biotechnol. 2015;42:1369–76.
148. Zhang K, Lian S, Shen X, Zhao X, Zhao W, Li C. Recombinant porcine
beta defensin 2 alleviates inflammatory responses induced by Escheri-
chia coli in IPEC-J2 cells. Int J Biol Macromol. 2022;208:890–900.
149. Shalovylo YI, Yusypovych YM, Hrunyk NI, Roman II, Zaika VK, Krynytskyy
HT, et al. Seed-derived defensins from Scots pine: structural and func‑
tional features. Planta. 2021;254:129.
150. Gaglione R, Dell’Olmo E, Bosso A, Chino M, Pane K, Ascione F, et al.
Novel human bioactive peptides identified in apolipoprotein B: evalua‑
tion of their therapeutic potential. Biochem Pharmacol. 2017;130:34–50.
151. Wang T, Wang Z, Mi J, Wang W, Li K, Qi X, et al. Recombinant avian
β-defensin produced by food-grade Lactococcus as a novel and potent
immunological enhancer adjuvant for avian vaccine. Probiotics Antimi‑
Ready to submit your research ? Choose BMC and benefit from:
crob Proteins. 2021;13:1833–46.
152. Thomas DS, Manoharan C, Rasalkar S, Mishra RK, Gopalapillai R. Recom‑
• fast, convenient online submission
binant expression of sericin-cecropin fusion protein and its functional
activity. Biotechnol Lett. 2020;42:1673–82. • thorough peer review by experienced researchers in your field
153. Orrapin S, Intorasoot A, Roytrakul S, Dechsupa N, Kantapan J, Onphat • rapid publication on acceptance
Y, et al. A novel recombinant javanicin with dual antifungal and anti-
• support for research data, including large and complex data types
proliferative activities. Sci Rep. 2019;9:18417.
154. Almasia NI, Molinari MP, Maroniche GA, Nahirñak V, Barrios Barón MP, • gold Open Access which fosters wider collaboration and increased citations
Taboga OA, et al. Successful production of the potato antimicrobial • maximum visibility for your research: over 100M website views per year
peptide Snakin-1 in baculovirus-infected insect cells and development
of specific antibodies. BMC Biotechnol. 2017;17:75. At BMC, research is always in progress.
155. Zhang L, Li G, Zhan N, Sun T, Cheng B, Li Y, et al. Expression of a Pseu-
domonas aeruginosa-targeted antimicrobial peptide T9W in Bacillus sub- Learn more biomedcentral.com/submissions
tilis using a maltose-inducible vector. Process Biochem. 2019;81:22–7.

Content courtesy of Springer Nature, terms of use apply. Rights reserved.


Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:

1. use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
2. use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
3. falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
4. use bots or other automated methods to access the content or redirect messages
5. override any security feature or exclusionary protocol; or
6. share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at

onlineservice@springernature.com

You might also like